Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Sci Rep ; 11(1): 18015, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34504172

ABSTRACT

Obesity and high-fat diet (HFD) consumption result in hypothalamic inflammation and metabolic dysfunction. While the TLR4 activation by dietary fats is a well-characterized pathway involved in the neuronal and glial inflammation, the role of its accessory proteins in diet-induced hypothalamic inflammation remains unknown. Here, we demonstrate that the knockdown of TLR4-interactor with leucine-rich repeats (Tril), a functional component of TLR4, resulted in reduced hypothalamic inflammation, increased whole-body energy expenditure, improved the systemic glucose tolerance and protection from diet-induced obesity. The POMC-specific knockdown of Tril resulted in decreased body fat, decreased white adipose tissue inflammation and a trend toward increased leptin signaling in POMC neurons. Thus, Tril was identified as a new component of the complex mechanisms that promote hypothalamic dysfunction in experimental obesity and its inhibition in the hypothalamus may represent a novel target for obesity treatment.


Subject(s)
Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Neurons/metabolism , Obesity/etiology , Pro-Opiomelanocortin/genetics , Toll-Like Receptor 4/genetics , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Diet, High-Fat/adverse effects , Energy Metabolism/genetics , Gene Expression Regulation , Glucose Tolerance Test , Hypothalamus/pathology , Inflammation , Intercellular Signaling Peptides and Proteins/deficiency , Male , Membrane Proteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/pathology , Obesity/metabolism , Obesity/pathology , Pro-Opiomelanocortin/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Toll-Like Receptor 4/metabolism
2.
Trials ; 22(1): 71, 2021 Jan 20.
Article in English | MEDLINE | ID: mdl-33472675

ABSTRACT

BACKGROUND: SARS-CoV-2, the virus that causes COVID-19, enters the cells through a mechanism dependent on its binding to angiotensin-converting enzyme 2 (ACE2), a protein highly expressed in the lungs. The putative viral-induced inhibition of ACE2 could result in the defective degradation of bradykinin, a potent inflammatory substance. We hypothesize that increased bradykinin in the lungs is an important mechanism driving the development of pneumonia and respiratory failure in COVID-19. METHODS: This is a phase II, single-center, three-armed parallel-group, open-label, active control superiority randomized clinical trial. One hundred eighty eligible patients will be randomly assigned in a 1:1:1 ratio to receive either the inhibitor of C1e/kallikrein 20 U/kg intravenously on day 1 and day 4 plus standard care; or icatibant 30 mg subcutaneously, three doses/day for 4 days plus standard care; or standard care alone, as recommended in the clinical trials published to date, which includes supplemental oxygen, non-invasive and invasive ventilation, antibiotic agents, anti-inflammatory agents, prophylactic antithrombotic therapy, vasopressor support, and renal replacement therapy. DISCUSSION: Accumulation of bradykinin in the lungs is a common side effect of ACE inhibitors leading to cough. In animal models, the inactivation of ACE2 leads to severe acute pneumonitis in response to lipopolysaccharide (LPS), and the inhibition of bradykinin almost completely restores the lung structure. We believe that inhibition of bradykinin in severe COVID-19 patients could reduce the lung inflammatory response, impacting positively on the severity of disease and mortality rates. TRIAL REGISTRATION: Brazilian Clinical Trials Registry Universal Trial Number (UTN) U1111-1250-1843. Registered on May/5/2020.


Subject(s)
Bradykinin/analogs & derivatives , COVID-19 Drug Treatment , Complement C1 Inhibitor Protein/administration & dosage , Respiratory Insufficiency/drug therapy , Adult , Angiotensin-Converting Enzyme 2/metabolism , Bradykinin/administration & dosage , Bradykinin/adverse effects , Bradykinin/antagonists & inhibitors , Bradykinin/immunology , Bradykinin/metabolism , Bradykinin B2 Receptor Antagonists/administration & dosage , Bradykinin B2 Receptor Antagonists/adverse effects , Brazil , COVID-19/complications , COVID-19/immunology , COVID-19/virology , Clinical Trials, Phase II as Topic , Complement C1 Inhibitor Protein/adverse effects , Drug Administration Schedule , Drug Therapy, Combination/adverse effects , Drug Therapy, Combination/methods , Humans , Injections, Intravenous , Injections, Subcutaneous , Kallikreins/antagonists & inhibitors , Kallikreins/metabolism , Randomized Controlled Trials as Topic , Respiratory Insufficiency/immunology , Respiratory Insufficiency/virology , SARS-CoV-2/isolation & purification , SARS-CoV-2/pathogenicity , Severity of Illness Index , Treatment Outcome
3.
J Clin Endocrinol Metab ; 106(2): 472-484, 2021 01 23.
Article in English | MEDLINE | ID: mdl-33180910

ABSTRACT

BACKGROUND: The brown adipose tissue (BAT) is a potential target for the treatment of obesity and metabolic disorders. Its activation by cold exposure or adrenergic drugs can increase systemic insulin sensitivity and improve lipid metabolism; however, little is known about the effects of specific dietary components on BAT activity. OBJECTIVES: We asked if a short-term (4 weeks) dietary intervention with olive oil could modify BAT activity in lean and overweight/obese volunteers. DESIGN: This was a 4-week open clinical trial in which all participants underwent a dietary intervention with extra-virgin olive oil supplementation. As the initial intake of olive oil was controlled all the participants were controls of themselves. RESULTS: The intervention resulted in significant increase in blood monounsaturated fatty acid levels, which was accompanied by increased BAT activity in lean but not in overweight/obese volunteers. In the lean group, an increase in leptin was detected after the intervention, and low leptin values at the beginning of the study were predictive of greater BAT activity after intervention. In addition, increase in leptin concentration was associated with increased BAT activity. Three known endogenous mediators of BAT activity, secretin, fibroblast growth factor 21 (FGF21), and 12,13-dihydroxy-9Z-octadecenoic acid (12,13-diHOME) were increased by intervention in lean, whereas only secretin and FGF21 were increased in subjects with excessive weight. CONCLUSION: This study provides clinical evidence for the impact of monounsaturated fatty acids on BAT activity and an advance in the understanding of the beneficial health effects of olive oil.


Subject(s)
Adipose Tissue, Brown/physiology , Obesity/diet therapy , Olive Oil/administration & dosage , Overweight/diet therapy , Thinness/diet therapy , Adipose Tissue, Brown/drug effects , Adult , Female , Follow-Up Studies , Humans , Male , Obesity/metabolism , Overweight/metabolism , Prognosis , Thinness/metabolism
4.
Int J Obes (Lond) ; 44(1): 178-185, 2020 01.
Article in English | MEDLINE | ID: mdl-31201362

ABSTRACT

BACKGROUND/OBJECTIVES: Hypothalamic neurons play a major role in the control of body mass. Obese subjects present radiologic signs of gliosis in the hypothalamus, which may reflect the damage or loss of neurons involved in whole-body energy homeostasis. It is currently unknown if hypothalamic gliosis (1) differs between obese nondiabetic (ND) and obese diabetic subjects (T2D) or (2) is modified by extensive body mass reduction via Roux-n-Y gastric bypass (RYGB). SUBJECTS/METHODS: Fifty-five subjects (all female) including lean controls (CT; n = 13), ND (n = 28), and T2D (n = 14) completed at least one study visit. Subjects underwent anthropometrics and a multi-echo MRI sequence to measure mean bilateral T2 relaxation time in the mediobasal hypothalamus (MBH) and two reference regions (amygdala and putamen). The obese groups underwent RYGB and were re-evaluated 9 months later. Analyses were by linear mixed models. RESULTS: Analyses of T2 relaxation time at baseline showed a group by region interaction only in the MBH (P < 0.0001). T2D had longer T2 relaxation times compared to either CT or ND groups. To examine the effects of RYGB on hypothalamic gliosis a three-way (group by region by time) mixed effects model adjusted for age was executed. Group by region (P < 0.0001) and region by time (P = 0.0005) interactions were significant. There was a reduction in MBH relaxation time by RYGB, and, although the T2D group still had higher T2 relaxation time overall compared to the ND group, the T2D group had significantly lower T2 relaxation time after surgery and the ND group showed a trend. The degree of reduction in MBH T2 relaxation time by RYGB was unrelated to clinical outcomes. CONCLUSION: T2 relaxation times, a marker of hypothalamic gliosis, are higher in obese women with T2D and are reduced by RYGB-induced weight loss.


Subject(s)
Bariatric Surgery , Diabetes Mellitus, Type 2/complications , Gliosis , Hypothalamus , Obesity , Female , Gliosis/diagnostic imaging , Gliosis/pathology , Humans , Hypothalamus/diagnostic imaging , Hypothalamus/pathology , Magnetic Resonance Imaging , Obesity/complications , Obesity/surgery , Treatment Outcome
5.
Acta Diabetol ; 56(12): 1333-1339, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31506721

ABSTRACT

AIMS: This study aimed to evaluate the effect of pioglitazone on brown adipose tissue function and hypothalamic gliosis in humans. Brown adipose tissue and the hypothalamus are regarded as important potential pharmacological targets to metabolic diseases, and defining the impact of current therapies on their structure and/or function could provide therapeutic advance in this field. METHODS: Six patients with type 2 diabetes were treated for 24 weeks with pioglitazone 30 mg/day as an add-on therapy. Brown adipose tissue glucose uptake and volume were determined using 18F-FDG PET/CT scans; hypothalamic gliosis was determined using MRI scans; blood was collected for hormone and biochemistry measurements. All tests were performed at inclusion and six months after pioglitazone introduction. RESULTS: Pioglitazone treatment led to a significant 3% body mass increase. There were neither changes in cold-induced brown adipose tissue glucose uptake and volume nor changes in hypothalamic gliosis. CONCLUSIONS: This is a proof-of-concept study that provides clinical evidence for a lack of action of a thiazolidinedione, pioglitazone, to promote homogeneous and measurable changes in brown adipose tissue volume and also in hypothalamic gliosis after 6 months of treatment.


Subject(s)
Adipose Tissue, Brown/drug effects , Diabetes Mellitus, Type 2/drug therapy , Gliosis/prevention & control , Hypothalamus/drug effects , Hypothalamus/pathology , Pioglitazone/pharmacology , Adipose Tissue, Brown/diagnostic imaging , Adipose Tissue, Brown/pathology , Adult , Aged , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/pathology , Drug Therapy, Combination , Female , Fluorodeoxyglucose F18 , Gliosis/diagnosis , Gliosis/pathology , Humans , Hypothalamus/diagnostic imaging , Male , Middle Aged , Obesity/complications , Obesity/diagnosis , Obesity/drug therapy , Obesity/pathology , Organ Size/drug effects , Overweight/complications , Overweight/diagnosis , Overweight/drug therapy , Overweight/pathology , Pioglitazone/administration & dosage , Positron Emission Tomography Computed Tomography , Proof of Concept Study , Thiazolidinediones/administration & dosage , Thiazolidinediones/pharmacology
6.
EBioMedicine ; 39: 448-460, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30522931

ABSTRACT

BACKGROUND: The Iroquois homeobox 3 (Irx3) gene has been identified as a functional long-range target of obesity-associated variants within the fat mass and obesity-associated protein (FTO) gene. It is highly expressed in the hypothalamus, and both whole-body knockout and hypothalamic restricted abrogation of its expression results in a lean phenotype, which is mostly explained by the resulting increased energy expenditure in the brown adipose tissue. Because of its potential implication in the pathogenesis of obesity, we evaluated the hypothalamic cell distribution of Irx3 and the outcomes of inhibiting its expression in a rodent model of diet-induced obesity. METHODS: Bioinformatics tools were used to evaluate the correlations between hypothalamic Irx3 and neurotransmitters, markers of thermogenesis and obesity related phenotypes. Droplet-sequencing analysis in >20,000 hypothalamic cells was used to explore the types of hypothalamic cells expressing Irx3. Lentivirus was used to inhibit hypothalamic Irx3 and the resulting phenotype was studied. FINDINGS: IRX3 is expressed predominantly in POMC neurons. Its expression is inhibited during prolonged fasting, as well as when mice are fed a high-fat diet. The partial inhibition of hypothalamic Irx3 using a lentivirus resulted in increased diet-induced body mass gain and adiposity due to increased caloric intake and reduced energy expenditure. INTERPRETATION: Contrary to the results obtained when lean mice are submitted to complete inhibition of Irx3, partial inhibition of hypothalamic Irx3 in obese mice causes an exacerbation of the obese phenotype. These data suggest that at least some of the Irx3 functions in the hypothalamus are regulated according to a hormetic pattern, and modulation of its expression can be a novel approach to modifying the body's energy-handling regulation. FUND: Sao Paulo Research Foundation grants 2013/07607-8 (LAV) and 2017/02983-2 (JDJ); NIH grants R01DK083567 (YBK).


Subject(s)
Diet, High-Fat/adverse effects , Down-Regulation , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Hypothalamus/metabolism , Obesity/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Body Weight , Cell Line , Computational Biology/methods , Disease Models, Animal , Energy Intake , Energy Metabolism , Fasting/metabolism , Humans , Male , Mice , Obesity/chemically induced , Obesity/metabolism , Phenotype , Sequence Analysis, RNA
7.
EBioMedicine ; 39: 436-447, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30502051

ABSTRACT

BACKGROUND: Inflammation is the most relevant mechanism linking obesity with insulin-resistance and metabolic disease. It impacts the structure and function of tissues and organs involved in metabolism, such as the liver, pancreatic islets and the hypothalamus. Brown adipose tissue has emerged as an important component of whole body energy homeostasis, controlling caloric expenditure through the regulation of non-shivering thermogenesis. However, little is known about the impact of systemic inflammation on the structure and function of brown adipose tissue. METHODS: The relations between IL10 and mitochondria structure/function and also with thermogenesis were evaluated by bioinformatics using human and rodent data. Real-time PCR, immunoblot, fluorescence and transmission electron microscopy were employed to determine the effect of IL10 in the brown adipose tissue of wild type and IL10 knockout mice. FINDINGS: IL10 knockout mice, a model of systemic inflammation, present severe structural abnormalities of brown adipose tissue mitochondria, which are round-shaped with loss of cristae structure and increased fragmentation. IL10 deficiency leads to newborn cold intolerance and impaired UCP1-dependent brown adipose tissue mitochondrial respiration. The reduction of systemic inflammation with an anti-TNFα monoclonal antibody partially rescued the structural but not the functional abnormalities of brown adipose tissue mitochondria. Using bioinformatics analyses we show that in both humans and mice, IL10 transcripts correlate with mitochondrial lipid metabolism and caspase gene expression. INTERPRETATION: IL10 and systemic inflammation play a central role in the regulation of brown adipose tissue by controlling mitochondrial structure and function. FUND: Sao Paulo Research Foundation grant 2013/07607-8.


Subject(s)
Adipose Tissue, Brown/cytology , Inflammation/pathology , Interleukin-10/genetics , Mitochondria/pathology , Shivering/genetics , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/pathology , Animals , Caspases/genetics , Cell Line , Cold Temperature , Computational Biology/methods , Energy Metabolism , Gene Knockout Techniques , Humans , Inflammation/genetics , Inflammation/metabolism , Lipid Metabolism , Male , Mice , Mitochondria/genetics , Mitochondria/metabolism , Uncoupling Protein 1/metabolism
8.
J Cell Physiol ; 233(12): 9426-9436, 2018 12.
Article in English | MEDLINE | ID: mdl-30063084

ABSTRACT

Hypothalamic sphingosine-1-phosphate receptor 1 (S1PR1), the G protein-coupled receptor 1 of sphingosine-1-phosphate, has been described as a modulator in the control of energy homeostasis in rodents. However, this mechanism is still unclear. Here, we evaluate the role of interleukin 6 (IL-6) associated with acute physical exercise in the control of the hypothalamic S1PR1-signal transducer and activator of transcription 3 (STAT3) axis. Acute exercise session and an intracerebroventricular IL-6 injection increased S1PR1 protein content and STAT3 phosphorylation in the hypothalamus of lean and obese mice accompanied by a reduction in food consumption. Transcriptome analysis indicated a strong positive correlation between Il-6 and S1pr1 messenger RNA in several tissues of genetically diverse BXD mice strains and humans, including in the hypothalamus. Interestingly, exercise failed to stimulate the S1PR1-STAT3 axis in IL-6 knockout mice and the disruption of hypothalamic-specific IL-6 action blocked the anorexigenic effects of exercise. Taken together, our results indicate that physical exercise modulates the S1PR1 protein content in the hypothalamus, through the central action of IL-6.


Subject(s)
Hypothalamus/metabolism , Interleukin-6/metabolism , Physical Conditioning, Animal , Receptors, Lysosphingolipid/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Animals , Humans , Injections, Intraventricular , Interleukin-6/administration & dosage , Interleukin-6/genetics , Male , Mice, Inbred C57BL , Mice, Obese , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Lysosphingolipid/genetics , Sphingosine-1-Phosphate Receptors
9.
Brain Behav Immun ; 73: 550-561, 2018 10.
Article in English | MEDLINE | ID: mdl-29935943

ABSTRACT

Hypothalamic hypoxia-inducible factor-1 (HIF-1) can regulate whole-body energy homeostasis in response to changes in blood glucose, suggesting that it acts as a sensor for systemic energy stores. Here, we hypothesized that hypothalamic HIF-1 could be affected by diet-induced obesity (DIO). We used eight-week old, male C57Bl6 mice, fed normal chow diet or with high fat diet for 1, 3, 7, 14 and 28 days. The expression of HIF-1alpha and HIF-1beta was measured by PCR and western blotting and its hypothalamic distribution was evaluated by fluorescence microscopy. Inhibition of HIF-1beta in arcuate nucleus of hypothalamus was performed using stereotaxic injection of shRNA lentiviral particles and animals were grouped under normal chow diet or high fat diet for 14 days. Using bioinformatics, we show that in humans, the levels of HIF-1 transcripts are directly correlated with those of hypothalamic transcripts for proteins involved in inflammation, regulation of apoptosis, autophagy, and the ubiquitin/proteasome system; furthermore, in rodents, hypothalamic HIF-1 expression is directly correlated with the phenotype of increased energy expenditure. In mice, DIO was accompanied by increased HIF-1 expression. The inhibition of hypothalamic HIF-1 by injection of an shRNA resulted in a further increase in body mass, a decreased basal metabolic rate, increased hypothalamic inflammation, and glucose intolerance. Thus, hypothalamic HIF-1 is increased during DIO, and its inhibition worsens the obesity-associated metabolic phenotype. Thus, hypothalamic HIF-1 emerges as a target for therapeutic intervention against obesity.


Subject(s)
Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Obesity/metabolism , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator/physiology , Blood Glucose/metabolism , Body Weight , Diet, High-Fat/adverse effects , Down-Regulation , Energy Metabolism , Hypothalamus/drug effects , Hypothalamus/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/physiopathology
10.
Metabolism ; 85: 171-182, 2018 08.
Article in English | MEDLINE | ID: mdl-29660453

ABSTRACT

BACKGROUND: The consumption of large amounts of dietary fats induces hypothalamic inflammation and impairs the function of the melanocortin system, leading to a defective regulation of caloric intake and whole-body energy expenditure. In mice fed a high-fat diet (HFD), TGF-ß1 expression was increased and NF-κB signaling was activated in proopiomelanocortin neurons, which plays an important role in the obesity-associated hypothalamic inflammation scenario. However, whether excessive hypothalamic TGF-ß1 impairs energy homeostasis remains unclear. OBJECTIVES: We aimed to investigate the role of diet-induced hypothalamic TGF-ß1 on inflammation and whole-body energy homeostasis. METHODS: A TGF-ß1 inhibitory lentiviral shRNA particle was stereotaxically injected bilaterally in the arcuate nucleus (ARC) of C57BL/6 mice fed a HFD. We assessed changes in body mass and adiposity, food intake, inflammatory markers, and the function of energy and glucose metabolism. RESULTS: TGF-ß1 down-regulation in the ARC-attenuated body-mass gain, reduced fat-mass accumulation, decreased hypothalamic inflammatory markers, and protected against HFD-induced lipohypertrophy of brown adipose tissue. In addition, the inhibition of hypothalamic TGF-ß1 increased the locomotor activity and improved whole-body lipid metabolism, which attenuated hepatic fat accumulation and serum triglyceride levels. No changes were observed in food intake and glucose homeostasis. CONCLUSION: Hypothalamic TGF-ß1 down-regulation attenuates hypothalamic inflammation and improves energy metabolism, resulting in lower body-mass gain and lower fat-mass accumulation, which protects mice from the development of obesity. Our data suggest that modulation of hypothalamic TGF-ß1 expression might be an effective strategy to treat obesity.


Subject(s)
Diet, High-Fat/adverse effects , Hypothalamus/metabolism , Inflammation/metabolism , Obesity/metabolism , Transforming Growth Factor beta1/metabolism , Adiposity/physiology , Animals , Down-Regulation , Eating/physiology , Energy Metabolism/physiology , Inflammation/genetics , Insulin Resistance/physiology , Male , Mice , Obesity/etiology , Obesity/genetics , Oxygen Consumption/physiology , RNA, Small Interfering , Transforming Growth Factor beta1/genetics
11.
Clin Endocrinol (Oxf) ; 88(3): 388-396, 2018 03.
Article in English | MEDLINE | ID: mdl-29280189

ABSTRACT

BACKGROUND: The combinations of adipokines and body mass parameters to estimate carotid atherosclerotic disease have not been completely delineated. OBJECTIVE: To test the combinations of well-established, easily accessible body mass indices and circulating biomarkers to identify increased carotid intima-media thickness (cIMT) in a primary prevention setting. DESIGN AND PATIENTS: In a cross-sectional analysis of 339 asymptomatic individuals with no history of cardiovascular events, inflammatory and insulin sensitivity biomarkers as well as adipokine levels were measured and combined with body mass parameters to evaluate the best marker for increased cIMT. RESULTS: As isolated parameters, body mass index (BMI) and adiponectin best identified abnormal cIMT (P = .04). Adiponectin levels were also linked to the relationship between BMI and cIMT (ß = 0.0371; P = .01). Twenty-nine individuals with increased cIMT were missed by BMI alone but detected by combining BMI and adiponectin measurements. When compared with BMI alone, the combination of adiponectin plus BMI improved the c-statistic (0.549-0.567) and the integrated discrimination improvement index (0.01725; P = .021). Segregation of individuals by the combined use of BMI + adiponectin is associated with significant differences in insulin sensitivity, glomerular filtration rate, systemic inflammatory activity, dyslipidaemia and cIMT. CONCLUSIONS: Combining plasma adiponectin measurements and BMI improves estimation of cIMT as compared to anthropometric parameters.


Subject(s)
Adiponectin/blood , Atherosclerosis/diagnosis , Body Mass Index , Carotid Intima-Media Thickness , Adult , Anthropometry , Biomarkers/blood , Cross-Sectional Studies , Female , Humans , Male , Risk Assessment
12.
Sci Rep ; 6: 29290, 2016 07 04.
Article in English | MEDLINE | ID: mdl-27373214

ABSTRACT

Obesity is the result of a long-term positive energy balance in which caloric intake overrides energy expenditure. This anabolic state results from the defective activity of hypothalamic neurons involved in the sensing and response to adiposity. However, it is currently unknown what the earliest obesity-linked hypothalamic defect is and how it orchestrates the energy imbalance present in obesity. Using an outbred model of diet-induced obesity we show that defective regulation of hypothalamic POMC is the earliest marker distinguishing obesity-prone from obesity-resistant mice. The early inhibition of hypothalamic POMC was sufficient to transform obesity-resistant in obesity-prone mice. In addition, the post-prandial change in the blood level of ß-endorphin, a POMC-derived peptide, correlates with body mass gain in rodents and humans. Taken together, these results suggest that defective regulation of POMC expression, which leads to a change of ß-endorphin levels, is the earliest hypothalamic defect leading to obesity.


Subject(s)
Hypothalamus/metabolism , Inflammation/metabolism , Obesity/metabolism , Pro-Opiomelanocortin/metabolism , beta-Endorphin/metabolism , Adolescent , Adult , Animals , Diet , Dietary Fats/metabolism , Energy Intake , Humans , Hypothalamus/immunology , Inflammation/immunology , Male , Mice , Mice, Obese , Obesity/immunology , Pro-Opiomelanocortin/immunology , Rats , Rats, Wistar , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...